The ARL6IP5 antibody targets the ADP-ribosylation factor-like 6 interacting protein 5 (ARL6IP5), a protein implicated in intracellular protein transport, oxidative stress response, and DNA repair. This antibody is used in research to quantify ARL6IP5 expression levels in tissues and cell lines, particularly in studies investigating chemoresistance mechanisms in cancers like ovarian carcinoma .
| Experimental Condition | Cellular Effects | Impact on Cisplatin Resistance |
|---|---|---|
| Overexpression (OE) | ↓ Proliferation, invasion, migration; ↑ apoptosis | Reduced resistance in OC and cisplatin-resistant (CisR) cells |
| Knockdown (KD) | ↑ Proliferation, invasion, migration; ↓ apoptosis | Increased resistance in both cell types |
Mechanistic Insights:
| Agent | Apoptotic Efficacy in OC Cells | Apoptotic Efficacy in CisR Cells |
|---|---|---|
| Cisplatin | Moderate | Low |
| Olaparib | Moderate | Low |
| rARL6IP5 | High | High |
Therapeutic Potential: rARL6IP5 demonstrated superior apoptotic activity compared to conventional agents, suggesting its utility as a novel chemotherapeutic .
ARL6IP5 (ADP-ribosylation-like factor 6 interacting protein 5) is a multifunctional protein also known as JWA in humans, Addicsin in mice, and GTRAP 3-18 or JM4 in rats. It belongs to the PRAF3 family with a large prenylated acceptor domain 1, primarily involved in intracellular protein trafficking .
To study ARL6IP5 function, researchers typically employ:
Genetic manipulation: Overexpression (OE) and knockdown (KD) studies using plasmid transfection. For example, transfection with 1 μg ARL6IP5 plasmid mixed with 50 μL serum-free media and 3 µL Lipofectamine 2000, followed by 48-hour incubation .
Protein interaction assays: Co-immunoprecipitation to identify binding partners (e.g., ATG12 interaction) .
Subcellular localization: Immunofluorescence microscopy using anti-ARL6IP5 antibodies (typically at 0.25-2 μg/mL concentration) .
Functional assays: Measurement of autophagy (LC3BII levels), apoptosis (TUNEL assay), and cellular proliferation in response to ARL6IP5 modulation .
For optimal Western blot results with ARL6IP5 antibodies:
Sample preparation: Extract proteins using RIPA buffer with protease inhibitors
Protein loading: 20-30 μg total protein per lane
Antibody concentration:
ARL6IP5 has a calculated molecular weight of 22kDa , so use appropriate percentage gels (12-15%)
Some antibodies detect multiple bands (22kDa and 48kDa) , so validation with positive controls is essential
For difficult samples, try HBAM buffer systems which have shown better results than enhanced buffers
Transfer time: 60-90 minutes at 100V for efficient protein transfer
When selecting an anti-ARL6IP5 antibody for immunohistochemistry (IHC):
Epitope specificity: Choose antibodies targeting well-conserved regions. The C-terminal region (sequence: NRLTDYISKVKE) has proven effective for generating specific antibodies .
Validation status: Select antibodies with documented IHC validation. For example, Atlas Antibodies' HPA015540 has been validated through the Human Protein Atlas project .
Species reactivity: Ensure compatibility with your experimental model. Available antibodies show reactivity to:
Staining protocol optimization:
Semi-quantitative scoring method for ARL6IP5 expression:
| Score | Intensity |
|---|---|
| 0 | Negative |
| 1 | Weak |
| 2 | Moderate |
| 3 | Strong |
This scoring system has been successfully employed in clinical studies correlating ARL6IP5 expression with patient outcomes .
To investigate ARL6IP5's role in autophagy and neurodegenerative diseases, consider this comprehensive experimental approach:
Establish relevant cellular models:
Manipulate ARL6IP5 expression:
Autophagy assessment:
α-synuclein aggregate measurement:
A11 antibody reactivity to measure toxic aggregates
GFP fluorescence quantification in A53T α-synuclein-GFP expressing cells
Measurement of LDH release to assess cellular toxicity
Evaluate the ARL6IP5/Rab1/ATG12 axis through co-immunoprecipitation
Assess subcellular localization changes of ARL6IP5 during autophagy induction
Determine the effects of autophagy inhibitors on ARL6IP5-mediated neuroprotection
Research has shown that ARL6IP5 overexpression increases autophagy by 150±54% compared to control, and reduces A53T α-synuclein fluorescence from 58±24 in control cells to 28±33 in ARL6IP5-transfected cells (p<0.0001) .
To investigate ARL6IP5's role in cisplatin resistance and DNA repair:
Establish cisplatin-resistant (CisR) cell lines:
Expose cancer cells (e.g., ovarian cancer lines OV90, SKOV3) to increasing concentrations of cisplatin
Validate resistance through IC50 determination
Manipulate ARL6IP5 expression:
Overexpression: Transfect with ARL6IP5 plasmid
Knockdown: Use siRNA targeting ARL6IP5
Treatment with recombinant ARL6IP5 protein (rARL6IP5)
Cell viability and apoptosis assays:
MTT/CCK-8 assays to determine cisplatin sensitivity
TUNEL assays to quantify apoptosis
Annexin V/PI staining followed by flow cytometry
Evaluate DNA repair proteins expression:
Western blot for XRCC1, PARP1, and other DNA repair proteins
Immunofluorescence for repair foci formation following cisplatin treatment
Comet assay to measure DNA damage and repair kinetics
Functional repair assays:
Host cell reactivation assays
Homologous recombination reporter assays
Non-homologous end joining assays
Compare efficacy of cisplatin, olaparib, and rARL6IP5 alone and in combination:
| Treatment | OV90 Concentration | SKOV3 Concentration |
|---|---|---|
| Cisplatin | 16.75 μM | 20 μM |
| Olaparib | 32.68 μM | 25 μM |
| rARL6IP5 | 1 μg/mL | 2.5 μg/mL |
Research has demonstrated that rARL6IP5 had greater apoptotic efficacy than cisplatin or olaparib in both cisplatin-sensitive and cisplatin-resistant ovarian cancer cells. Notably, while cisplatin and olaparib lost efficacy in resistant cells, rARL6IP5 maintained its apoptotic effect, suggesting it acts through pathways that remain functional despite cisplatin resistance .
To address contradictory findings about ARL6IP5 function across different cell types:
Conduct systematic analysis across multiple cell types:
Neuronal cells (SH-SY5Y, primary neurons)
Cancer cells (OV90, SKOV3, etc.)
Normal cells (fibroblasts, HEK293)
Baseline expression profiling:
Quantitative RT-PCR for ARL6IP5 mRNA levels
Western blot for protein expression using multiple validated antibodies
Single-cell RNA-seq to identify cell populations with varying expression
Identify cell-type specific binding partners:
Co-immunoprecipitation followed by mass spectrometry
Proximity labeling (BioID, APEX) to capture transient interactions
Yeast two-hybrid screening with cell-type specific cDNA libraries
Post-translational modification analysis:
Phosphorylation: Phospho-specific antibodies or phosphoproteomic analysis
Ubiquitination: Immunoprecipitation under denaturing conditions
Other modifications: Mass spectrometry-based approaches
Generate ARL6IP5 knockout in diverse cell lines using CRISPR-Cas9
Perform complementation with:
Wild-type ARL6IP5
Domain-specific mutants
Chimeric proteins with interacting partners
Autophagy context: Compare autophagy induction by ARL6IP5 in:
DNA repair context: Evaluate XRCC1/PARP1 interaction with ARL6IP5 in:
Cisplatin-sensitive vs. resistant cancer cells
Normal vs. cancer cells
To study the therapeutic potential of recombinant ARL6IP5 protein (rARL6IP5):
Expression system selection:
Bacterial (E. coli): Suitable for non-glycosylated versions
Mammalian (CHO cells): For properly folded protein with post-translational modifications
Insect cells (Sf9): Alternative for complex proteins
Purification strategy:
Affinity chromatography (His-tag or GST-tag)
Size exclusion chromatography for high purity
Endotoxin removal for in vivo applications
Quality assessment:
SDS-PAGE and Western blot
Circular dichroism for secondary structure
Activity assays to confirm functionality
Labeling strategies:
Fluorescent tagging (maintaining function)
Radioisotope labeling for in vivo tracking
Uptake mechanisms:
Flow cytometry to quantify cellular uptake
Confocal microscopy for intracellular localization
Endocytosis inhibitors to determine entry pathways
Cancer models:
Neurodegenerative models:
α-synuclein aggregation: Fluorescence measurement in GFP-A53T expressing cells
Autophagy induction: LC3B-II Western blot and puncta quantification
Neuroprotection assays: LDH release and cell viability measurements
Dose optimization:
Range-finding studies (based on in vitro EC50)
Administration route comparison (IV, IP, etc.)
Pharmacokinetic profiling
Disease models:
Xenograft models for cancer studies
α-synuclein transgenic mice for PD studies
Toxicity studies in multiple species
Research has shown that rARL6IP5 has significant therapeutic potential in cisplatin-resistant ovarian cancer, with greater apoptotic efficacy than conventional chemotherapeutics .
To elucidate how ARL6IP5 simultaneously regulates autophagy and DNA repair:
Domain mapping:
Generate truncation mutants to identify functional domains
Assess interaction with ATG12 (autophagy) vs. XRCC1/PARP1 (DNA repair)
Determine if interactions are mutually exclusive or can occur simultaneously
Structural determination:
X-ray crystallography of ARL6IP5 alone and in complexes
Cryo-EM for larger protein assemblies
NMR for dynamic interaction studies
Real-time imaging:
Live-cell imaging with fluorescently tagged ARL6IP5 and partners
FRET/BRET assays to measure protein-protein interactions in real-time
Photoactivation studies to track protein movement between compartments
Stress-induced relocalization:
Track ARL6IP5 localization after:
DNA damage (cisplatin treatment)
Autophagy induction (starvation, rapamycin)
Combined stressors
Quantify colocalization with organelle markers
Phosphoproteomics:
Global phosphorylation changes upon ARL6IP5 modulation
Identification of kinases/phosphatases regulating ARL6IP5
Mutational analysis of key phosphorylation sites
Transcriptional regulation:
RNA-seq after ARL6IP5 modulation
ChIP-seq to identify transcription factors regulated by ARL6IP5
Pathway enrichment analysis to identify coordinated gene programs
Neurodegenerative disease models:
Compare ARL6IP5 function in:
α-synuclein models (PD)
Amyloid-β models (AD)
SOD1 models (ALS)
Assess if autophagy predominates in these contexts
Cancer models:
Compare mechanism in cisplatin-sensitive vs. resistant cells
Determine if DNA repair functions predominate in cancer contexts
Assess if tumor microenvironment influences ARL6IP5 function
Experimental evidence indicates ARL6IP5 serves as a multifunctional protein capable of:
Binding ATG12 to promote autophagy and reduce α-synuclein aggregation
Suppressing DNA repair proteins XRCC1 and PARP1 to enhance cisplatin sensitivity
Understanding the molecular switch determining pathway selection would provide critical insights for therapeutic targeting.
When encountering inconsistent staining patterns with ARL6IP5 antibodies:
Cross-validation with multiple antibodies:
Compare polyclonal antibodies from different sources
Verify with antibodies targeting different epitopes
Include knockout/knockdown controls
Epitope masking assessment:
Test multiple antigen retrieval methods:
Heat-induced (citrate buffer, pH 6.0)
Enzymatic (proteinase K)
High pH (EDTA buffer, pH 9.0)
Extend retrieval times for formalin-fixed tissues
Fixation considerations:
Optimize fixation time (overfixation can mask epitopes)
Compare different fixatives (formalin vs. alcohol-based)
Prepare fresh frozen sections as alternative
Tissue processing:
Signal amplification:
Try polymer-based detection systems
Consider tyramide signal amplification for weak signals
Optimize incubation times and temperatures
Background reduction:
Include blocking steps (serum from secondary antibody species)
Add detergents (0.1-0.3% Triton X-100) to reduce non-specific binding
Use avidin/biotin blocking for tissues with endogenous biotin
Scoring system standardization:
Implement the validated semi-quantitative scoring method :
Have multiple observers score independently
Resolve discrepancies through consensus review
Include training sets for new observers to ensure consistency
To determine if ARL6IP5 directly or indirectly affects α-synuclein:
In vitro binding assays:
Pull-down assays with purified proteins
Surface plasmon resonance to measure binding kinetics
Isothermal titration calorimetry for thermodynamic parameters
Cellular colocalization:
Super-resolution microscopy (STED, STORM)
Proximity ligation assay to detect close association (<40nm)
FRET analysis between tagged proteins
Autophagy manipulation:
Use autophagy inhibitors (3-MA, bafilomycin A1) alongside ARL6IP5 overexpression
Compare wild-type ATG5/7 with autophagy-deficient cells
Quantify α-synuclein levels with immunoblotting and fluorescence
Selective autophagy markers:
Assess p62/SQSTM1 colocalization with α-synuclein
Evaluate ubiquitination status of α-synuclein
Determine if ARL6IP5 affects α-synuclein ubiquitination
Domain mapping:
Generate ARL6IP5 mutants lacking autophagy-inducing domains
Test if these mutants still affect α-synuclein levels
Map minimal region required for effect
Interactome comparison:
Identify proteins that interact with both ARL6IP5 and α-synuclein
Silence these mediators to determine if they're required for the effect
Reconstitute the pathway with purified components
Experimental evidence suggests that ARL6IP5 reduces α-synuclein burden by enhancing autophagy rather than direct interaction:
ARL6IP5 overexpression decreases A53T α-synuclein fluorescence by approximately 52% (from 58±24 to 28±33, p<0.0001)
ARL6IP5 knockdown increases α-synuclein toxicity by 15±7% (p=0.018)
ARL6IP5 increases autophagy marker LC3B-II by approximately 155±46% when co-expressed with α-synuclein
To integrate multi-omics for understanding tissue-specific ARL6IP5 functions:
Transcriptomics:
RNA-seq across tissues with differential ARL6IP5 expression
Single-cell RNA-seq to identify cell-type specific expression patterns
Alternative splicing analysis to detect tissue-specific isoforms
Proteomics:
Global proteome profiling after ARL6IP5 modulation
Interactome analysis in different tissues/cell types
Post-translational modification mapping
Metabolomics:
Untargeted metabolomics to identify affected pathways
Stable isotope tracing to track metabolic flux
Lipid profiling given ARL6IP5's membrane association
Network-based integration:
Construct protein-protein interaction networks
Pathway enrichment analysis across omics layers
Identify tissue-specific network modules
Machine learning approaches:
Predictive modeling of ARL6IP5 functions based on multi-omics signatures
Feature importance analysis to identify key regulatory nodes
Clustering to identify functionally similar tissues
Tissue-specific knockouts:
Generate conditional knockout models targeting specific tissues
Compare phenotypes across neural, cancer, and normal tissues
Conduct rescue experiments with tissue-specific promoters
3D organoid models:
Develop organoids from different tissues with ARL6IP5 modulation
Compare response to stressors (e.g., cisplatin, nutrient deprivation)
Assess tissue-specific pathways through targeted interventions
Human sample analysis:
Biomarker development:
Identify tissue-specific signatures associated with ARL6IP5 function
Develop predictive models for treatment response
Stratify patients based on multi-omics profiles
For studying ARL6IP5 in neuroinflammation contexts:
Microglia-neuron co-culture systems:
Primary cultures or iPSC-derived cells
3D spheroid co-cultures to model tissue architecture
Microfluidic platforms for controlled cell-cell interaction
Brain organoids:
Develop region-specific organoids (midbrain for PD studies)
Generate organoids with multiple cell types (neurons, astrocytes, microglia)
Gene editing to modulate ARL6IP5 expression in specific cell types
Cytokine profiling:
Multiplex cytokine arrays after ARL6IP5 modulation
Single-cell secretome analysis
In situ cytokine detection in tissue sections
Glial activation markers:
Flow cytometry for microglial/astrocyte activation markers
Live imaging of calcium signaling and morphological changes
Transcriptional profiling of inflammatory gene signatures
Spatial mapping techniques:
Visium spatial transcriptomics to map expression patterns
CODEX multiplexed protein imaging
MERFISH for single-cell spatial transcriptomics
Single-cell multi-omics:
CITE-seq for simultaneous protein and RNA profiling
Single-cell ATAC-seq to assess chromatin accessibility
Spatial proteomics with subcellular resolution
Disease-specific models:
α-synuclein preformed fibril (PFF) injection models
LPS-induced neuroinflammation
Transgenic models with cell-type specific ARL6IP5 modulation
Advanced imaging:
Two-photon imaging of glial dynamics in live animals
PET imaging with TSPO ligands for neuroinflammation
Multimodal imaging combining structural and functional readouts
Investigate if ARL6IP5's autophagy-promoting function (150±54% increase) affects:
Inflammasome activation and regulation
Clearance of protein aggregates triggering inflammation
Mitophagy of damaged mitochondria releasing DAMPs
Explore potential direct interactions with:
NF-κB pathway components
NLRP3 inflammasome proteins
Toll-like receptors and downstream mediators
To assess ARL6IP5 as a cancer prognostic biomarker:
Prognostic value assessment:
Kaplan-Meier survival analysis stratified by ARL6IP5 expression
Cox proportional hazards regression for multivariate analysis
Time-dependent ROC curve analysis for predictive accuracy
Cross-validation approaches:
Training/validation cohort design (70/30 split)
Leave-one-out cross-validation
External validation in independent cohorts
Multi-platform analysis:
Correlate protein expression with mRNA levels
Assess genomic alterations affecting ARL6IP5 (copy number, mutations)
Integrate with pathway activation signatures
Functional subtype identification:
Determine if ARL6IP5 has different prognostic value in:
Different molecular subtypes of each cancer
Patients with specific treatment histories
Tumors with varied DNA repair capacities
Circulating biomarker assessment:
Evaluate ARL6IP5 in circulating tumor cells
Assess ARL6IP5 in extracellular vesicles
Develop sensitive detection methods (e.g., digital ELISA)
Longitudinal monitoring:
Serial sampling during treatment and follow-up
Correlation with disease progression and treatment response
Integration with other established biomarkers
To reconcile contradictory findings about ARL6IP5 for translational development:
Structured comparison across studies:
Catalog experimental conditions (cell types, assays, endpoints)
Standardize effect sizes for cross-study comparison
Identify patterns related to specific experimental variables
Quality assessment framework:
Evaluate methodological rigor of contradictory studies
Assess antibody validation standards
Examine statistical approaches and sample sizes
Cellular context classification:
Normal vs. cancer cells
Neuronal vs. non-neuronal cells
Proliferating vs. differentiated cells
Stress context analysis:
Baseline vs. genotoxic stress (cisplatin)
Proteotoxic stress (protein aggregation)
Metabolic stress (nutrient deprivation)
Side-by-side comparison in standardized systems:
Test multiple functions simultaneously in the same cell type
Evaluate concentration-dependent effects
Assess temporal dynamics of different functions
Isoform-specific analysis:
Characterize expression of different splice variants across tissues
Test functional differences between isoforms
Develop isoform-specific detection methods
Therapeutic context definition:
Clearly define disease context (cancer type, neurodegeneration type)
Establish primary pathway of interest (autophagy, DNA repair)
Identify biomarkers for context-appropriate application
Risk mitigation strategies:
Develop companion diagnostics for appropriate patient selection
Design combination approaches addressing potential compensatory mechanisms
Establish monitoring protocols for expected and unexpected effects
Reconciling contradictory findings about ARL6IP5 is essential as research shows dual functions:
Promoting autophagy and reducing α-synuclein aggregation in neurodegenerative contexts
Suppressing DNA repair and enhancing cisplatin sensitivity in cancer contexts
These differences may represent true biological context-dependency rather than experimental artifacts, suggesting context-specific therapeutic applications.